Bcl-2-mediated cell survival promotes metastasis of EpH4 betaMEKDD mammary epithelial cells

Mol Cancer Res. 2004 Oct;2(10):551-6.

Abstract

The majority of patients who succumb to cancer die from metastatic disease progression rather than from the primary tumor. Elucidation of the mechanisms underlying tissue-specific metastasis is essential to the development of effective therapies. The mitogen-activated protein kinase kinase (MEK) pathway is frequently activated in human tumors and has been shown to regulate genes involved in proliferation, migration, and invasion. Studies with MEK-transformed EpH4 mouse mammary epithelial cells showed that these cells are highly tumorigenic but have a limited metastatic ability. Detachment of epithelial cells from the extracellular matrix causes disruption of the actin cytoskeleton and induces apoptosis. Several metastatic breast carcinoma cell lines have been shown to be resistant to cell death following actin disruption. This death-resistant phenotype can be modeled by overexpressing the antiapoptotic Bcl-2 protein in cells. This suggests that mechanisms that regulate survival of extravasated tumor cells may enhance metastatic efficiency. Therefore, we examined whether expression of Bcl-2 in MEK-transformed EpH4 mammary epithelial cells could provide a survival advantage and promote metastasis. Expression of Bcl-2 in parental EpH4 mammary epithelial cells or MEK-transformed cells was insufficient to induce increased migration, invasion, or tumor development. However, Bcl-2 expression markedly enhanced spontaneous lung metastasis from orthotopically implanted primary tumors. These results clearly show that mechanisms that regulate primary tumor development are distinct from those that promote metastasis and that assays designed to isolate genes involved in transformation may fail to identify genes that are critical regulators of metastasis.

MeSH terms

  • Animals
  • Breast Neoplasms / pathology*
  • Breast Neoplasms / physiopathology
  • Cell Line, Transformed
  • Cell Line, Tumor
  • Cell Survival / physiology
  • Epithelial Cells / cytology
  • Female
  • Gene Expression Regulation, Neoplastic / physiology
  • Humans
  • Lung Neoplasms / physiopathology
  • Lung Neoplasms / secondary*
  • Mammary Glands, Animal / cytology*
  • Mice
  • Mice, Inbred BALB C
  • Mitogen-Activated Protein Kinase Kinases / genetics
  • Neoplasm Metastasis / genetics*
  • Proto-Oncogene Proteins c-bcl-2 / genetics*
  • Proto-Oncogene Proteins c-bcl-2 / metabolism

Substances

  • Proto-Oncogene Proteins c-bcl-2
  • Mitogen-Activated Protein Kinase Kinases